1
|
Pan X, Wang X, Zhang Y, Yang X. Development of a PSMA and EphA2 dual-receptor-activated fluorescent probe for wash-free prostate cancer cell imaging. Bioorg Med Chem 2025; 125:118217. [PMID: 40315731 DOI: 10.1016/j.bmc.2025.118217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/03/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
Prostate cancer (PCa) remains a leading cause of cancer-related mortality in men, underscoring the need for improved diagnostic tools. In this study, a novel dual-receptor-activated fluorescence probe (9) designed to simultaneously bind to prostate-specific membrane antigen (PSMA) and erythropoietin-producing hepatocellular receptor type A2 (EphA2), two key biomarkers in PCa, was developed and applied for wash-free cell imaging. The probe was synthesized by conjugating both PSMA- and EphA2-targeting ligands with a sulfonated benzothiazole environment-sensitive dye. 9 demonstrated good binding affinity (Kd = 34 nM for PSMA, 9.70 nM for EphA2) with significant fluorescence activation upon receptor binding (14.1-fold for PSMA; 8.6-fold for EphA2). Cell studies demonstrated 9 showed negligible cytotoxicity and good targeting specificity, which could enable convenient monitoring of the dynamic biological processes in PCa, such as ligand-receptor interactions. The study may also provide insights into the design and applications of multiple-receptor-activated fluorescence probes.
Collapse
Affiliation(s)
- Xingcan Pan
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
| | - Xiao Wang
- Department of Nuclear Medicine, Peking University People's Hospital, Beijing 100044, China
| | - Yijing Zhang
- Department of Nuclear Medicine, Peking University People's Hospital, Beijing 100044, China
| | - Xing Yang
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China; Department of Nuclear Medicine, Peking University People's Hospital, Beijing 100044, China; Yunnan Baiyao Group, Kunming 650000, China.
| |
Collapse
|
2
|
Kumar P, Kumar A, Kumar V. Role of Microbiota-Derived Metabolites in Prostate Cancer Inflammation and Progression. Cell Biochem Funct 2025; 43:e70050. [PMID: 39891389 DOI: 10.1002/cbf.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/25/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Prostate cancer (PCa) is the most commonly detected malignancy in men worldwide. PCa is a slow-growing cancer with the absence of symptoms at early stages. The pathogenesis has not been entirely understood including the key risk factors related to PCa development like diet and microbiota derived metabolites. Microbiota may influence the host's immunological responses, inflammatory responses, and metabolic pathways, which may be crucial for the development and metastasis. Similarly, short-chain fatty acids, methylamines, hippurate, bile acids, and other metabolites generated by microbiota may have potential roles in cancer inflammation and progression of cancer. Most studies have focused on the role of metabolites and their pathways involved in chronic inflammation, tumor initiation, proliferation, and progression. In summary, the review discusses the role of microbiota and microbial-derived metabolite-built strategies in inflammation and progression of the PCa.
Collapse
Affiliation(s)
- Pradeep Kumar
- Department of NMR, All India Institute of Medical Sciences, New Delhi, India
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
| | - Virendra Kumar
- Department of NMR, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
3
|
Yang J, Wang H, Li B, Liu J, Zhang X, Wang Y, Peng J, Gao L, Wang X, Hu S, Zhang W, Hong L. Inhibition of ACSS2 triggers glycolysis inhibition and nuclear translocation to activate SIRT1/ATG5/ATG2B deacetylation axis, promoting autophagy and reducing malignancy and chemoresistance in ovarian cancer. Metabolism 2025; 162:156041. [PMID: 39362518 DOI: 10.1016/j.metabol.2024.156041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Metabolic reprogramming is a hallmark of cancer, characterized by a high dependence on glycolysis and an enhanced utilization of acetate as an alternative carbon source. ACSS2 is a critical regulator of acetate metabolism, playing a significant role in the development and progression of various malignancies. ACSS2 facilitates the conversion of acetate to acetyl-CoA, which participates in multiple metabolic pathways and functions as an epigenetic regulator of protein acetylation, thereby modulating key cellular processes such as autophagy. However, the roles and intrinsic connections of ACSS2, glycolysis, protein acetylation, and autophagy in ovarian cancer (OC) remain to be elucidated. BASIC PROCEDURES Utilizing clinical specimens and online databases, we analysed the expression of ACSS2 in OC and its relationship with clinical prognosis. By knocking down ACSS2, we evaluated its effects on the malignant phenotype, acetate metabolism, glycolysis, and autophagy. The metabolic alterations in OC cells were comprehensively analysed using Seahorse assays, transmission electron microscopy, membrane potential measurements, and stable-isotope labeling techniques. CUT&TAG and co-immunoprecipitation techniques were employed to explore the deacetylation of autophagy-related proteins mediated by ACSS2 via SIRT1. Additionally, through molecular docking, transcriptome sequencing, and metabolomics analyses, we validated the pharmacological effects of paeonol on ACSS2 and the glycolytic process in OC cells. Finally, both in vitro and in vivo experiments were performed to investigate the impact of paeonol on autophagy and its anti-OC effects mediated through the ACSS2/SIRT1 deacetylation axis. MAIN FINDINGS ACSS2 is significantly upregulated in OC and is associated with poor prognosis. Knockdown of ACSS2 inhibits OC cells proliferation, migration, invasion, angiogenesis, and platinum resistance, while reducing tumour burden in vivo. Mechanistically, inhibiting ACSS2 reduces acetate metabolism and suppresses glycolysis by targeting HXK2. This glycolytic reduction promotes the translocation of ACSS2 from the cytoplasm to the nucleus, leading to increased expression of the deacetylase SIRT1. SIRT1 mediates the deacetylation of autophagy-related proteins, such as ATG5 and ATG2B, thereby significantly activating autophagy in OC cells and exerting antitumor effects. Paeonol inhibits acetate metabolism and glycolysis in OC cells by targeting ACSS2. Paeonol activates autophagy through the ACSS2/SIRT1/ATG5/ATG2B deacetylation axis, demonstrating inhibition of OC in vitro and in vivo. PRINCIPAL CONCLUSIONS Pae can serve as an effective, low-toxicity, multi-targeted drug targeting ACSS2 and glycolysis. It activates autophagy through the ACSS2/SIRT1/ATG5/ATG2B deacetylation signalling cascade, thereby exerting anti-OC effects. Our study provides new insights into the malignant mechanisms of OC and offers a novel strategy for its treatment.
Collapse
Affiliation(s)
- Jiang Yang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China; Department of Obstetrics and Gynaecology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China
| | - Haoyu Wang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Bingshu Li
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Jingchun Liu
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Xiaoyi Zhang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Ying Wang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Jiaxin Peng
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Likun Gao
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Xinqi Wang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Siyuan Hu
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Wenyi Zhang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Li Hong
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China.
| |
Collapse
|
4
|
Wakui M, Tsuchiya J, Kase C, Yokoyama K, Yamamoto M, Tateishi U. Comparative Study of Metabolic Tumor Volume in Multiple Myeloma and Related Plasma Cell Dyscrasias: 11C-Acetate PET vs. 18F-FDG PET. Cureus 2024; 16:e71605. [PMID: 39421283 PMCID: PMC11485584 DOI: 10.7759/cureus.71605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Accurate diagnosis of multiple myeloma (MM) and related disorders depends on imaging studies for lesion detection, which is crucial for treatment planning. The 18F-fluorodeoxyglucose (FDG) PET imaging system is well-established, with high sensitivity and specificity in identifying myeloma lesions. Additionally, 11C-acetate serves as an effective radiotracer for detecting MM lesions. Metabolic tumor volume (MTV) measured with 18F-FDG PET has been suggested as a prognostic factor in MM patients. This study aimed to compare the feasibility of measuring MTV in patients with myeloma-related diseases using 11C-acetate or 18F-FDG PET/CT. Methods We retrospectively reviewed six patients with MM - three with symptomatic MM and three with smoldering MM - and one patient with a myeloma-related disorder, all of whom underwent both 11C-acetate and 18F-FDG PET/CT scans. Using a dedicated workstation (PET-STAT; AdIn Research, Inc., Tokyo, Japan) equipped with a standardized uptake value (SUV)-based automated contouring program, we calculated the SUV for MTV. Bone areas with an SUV above thresholds of 2.0 or 2.5 were grouped accordingly. Results MTV detection in the whole body was significantly higher with 11C-acetate compared to 18F-FDG at both thresholds. For the 2.5 threshold, MTV was 129 ± 109 mL versus 3.93 ± 9.38 mL (p = 0.016), and for the 2.0 threshold, it was 316 ± 221 mL versus 12.1 ± 23.7 mL (p = 0.016). Conclusions MTV measurements were significantly higher in PET/CT using 11C-acetate compared to 18F-FDG. This finding highlights the potential superiority of 11C-acetate over 18F-FDG for assessing the MTV of lesions in patients with MM.
Collapse
Affiliation(s)
- Momo Wakui
- Diagnostic Radiology and Nuclear Medicine, Institute of Science Tokyo, Tokyo, JPN
| | - Junichi Tsuchiya
- Diagnostic Radiology and Nuclear Medicine, Institute of Science Tokyo, Tokyo, JPN
| | - Chikara Kase
- Diagnostic Radiology and Nuclear Medicine, Institute of Science Tokyo, Tokyo, JPN
| | - Kota Yokoyama
- Diagnostic Radiology and Nuclear Medicine, Institute of Science Tokyo, Tokyo, JPN
| | | | - Ukihide Tateishi
- Diagnostic Radiology and Nuclear Medicine, Institute of Science Tokyo, Tokyo, JPN
| |
Collapse
|
5
|
Baniasadi A, Das JP, Prendergast CM, Beizavi Z, Ma HY, Jaber MY, Capaccione KM. Imaging at the nexus: how state of the art imaging techniques can enhance our understanding of cancer and fibrosis. J Transl Med 2024; 22:567. [PMID: 38872212 PMCID: PMC11177383 DOI: 10.1186/s12967-024-05379-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Both cancer and fibrosis are diseases involving dysregulation of cell signaling pathways resulting in an altered cellular microenvironment which ultimately leads to progression of the condition. The two disease entities share common molecular pathophysiology and recent research has illuminated the how each promotes the other. Multiple imaging techniques have been developed to aid in the early and accurate diagnosis of each disease, and given the commonalities between the pathophysiology of the conditions, advances in imaging one disease have opened new avenues to study the other. Here, we detail the most up-to-date advances in imaging techniques for each disease and how they have crossed over to improve detection and monitoring of the other. We explore techniques in positron emission tomography (PET), magnetic resonance imaging (MRI), second generation harmonic Imaging (SGHI), ultrasound (US), radiomics, and artificial intelligence (AI). A new diagnostic imaging tool in PET/computed tomography (CT) is the use of radiolabeled fibroblast activation protein inhibitor (FAPI). SGHI uses high-frequency sound waves to penetrate deeper into the tissue, providing a more detailed view of the tumor microenvironment. Artificial intelligence with the aid of advanced deep learning (DL) algorithms has been highly effective in training computer systems to diagnose and classify neoplastic lesions in multiple organs. Ultimately, advancing imaging techniques in cancer and fibrosis can lead to significantly more timely and accurate diagnoses of both diseases resulting in better patient outcomes.
Collapse
Affiliation(s)
- Alireza Baniasadi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA.
| | - Jeeban P Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Conor M Prendergast
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Zahra Beizavi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Hong Y Ma
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | | | - Kathleen M Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| |
Collapse
|
6
|
Pu Y, Li L, Peng H, Liu L, Heymann D, Robert C, Vallette F, Shen S. Drug-tolerant persister cells in cancer: the cutting edges and future directions. Nat Rev Clin Oncol 2023; 20:799-813. [PMID: 37749382 DOI: 10.1038/s41571-023-00815-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 09/27/2023]
Abstract
Drug-tolerant persister (DTP) cell populations were originally discovered in antibiotic-resistant bacterial biofilms. Similar populations with comparable features have since been identified among cancer cells and have been linked with treatment resistance that lacks an underlying genomic alteration. Research over the past decade has improved our understanding of the biological roles of DTP cells in cancer, although clinical knowledge of the role of these cells in treatment resistance remains limited. Nonetheless, targeting this population is anticipated to provide new treatment opportunities. In this Perspective, we aim to provide a clear definition of the DTP phenotype, discuss the underlying characteristics of these cells, their biomarkers and vulnerabilities, and encourage further research on DTP cells that might improve our understanding and enable the development of more effective anticancer therapies.
Collapse
Affiliation(s)
- Yi Pu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Li
- Lung Cancer Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Haoning Peng
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Lunxu Liu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Dominique Heymann
- Nantes Université, CNRS, UMR6286, US2B, Nantes, France
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Caroline Robert
- INSERM U981, Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - François Vallette
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France.
- Nantes Université, INSERM, U1307, CRCI2NA, Nantes, France.
| | - Shensi Shen
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Shegani A, Kealey S, Luzi F, Basagni F, Machado JDM, Ekici SD, Ferocino A, Gee AD, Bongarzone S. Radiosynthesis, Preclinical, and Clinical Positron Emission Tomography Studies of Carbon-11 Labeled Endogenous and Natural Exogenous Compounds. Chem Rev 2023; 123:105-229. [PMID: 36399832 PMCID: PMC9837829 DOI: 10.1021/acs.chemrev.2c00398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Indexed: 11/19/2022]
Abstract
The presence of positron emission tomography (PET) centers at most major hospitals worldwide, along with the improvement of PET scanner sensitivity and the introduction of total body PET systems, has increased the interest in the PET tracer development using the short-lived radionuclides carbon-11. In the last few decades, methodological improvements and fully automated modules have allowed the development of carbon-11 tracers for clinical use. Radiolabeling natural compounds with carbon-11 by substituting one of the backbone carbons with the radionuclide has provided important information on the biochemistry of the authentic compounds and increased the understanding of their in vivo behavior in healthy and diseased states. The number of endogenous and natural compounds essential for human life is staggering, ranging from simple alcohols to vitamins and peptides. This review collates all the carbon-11 radiolabeled endogenous and natural exogenous compounds synthesised to date, including essential information on their radiochemistry methodologies and preclinical and clinical studies in healthy subjects.
Collapse
Affiliation(s)
- Antonio Shegani
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Steven Kealey
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Federico Luzi
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Filippo Basagni
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum−University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Joana do Mar Machado
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Sevban Doğan Ekici
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Alessandra Ferocino
- Institute
of Organic Synthesis and Photoreactivity, Italian National Research Council, via Piero Gobetti 101, 40129 Bologna, Italy
| | - Antony D. Gee
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Salvatore Bongarzone
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, King’s Health Partners, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
8
|
Yang YF, Li CH, Cai HY, Lin BS, Kim CH, Chang YC. Application of Metabolic Reprogramming to Cancer Imaging and Diagnosis. Int J Mol Sci 2022; 23:15831. [PMID: 36555470 PMCID: PMC9782057 DOI: 10.3390/ijms232415831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Cellular metabolism governs the signaling that supports physiological mechanisms and homeostasis in an individual, including neuronal transmission, wound healing, and circadian clock manipulation. Various factors have been linked to abnormal metabolic reprogramming, including gene mutations, epigenetic modifications, altered protein epitopes, and their involvement in the development of disease, including cancer. The presence of multiple distinct hallmarks and the resulting cellular reprogramming process have gradually revealed that these metabolism-related molecules may be able to be used to track or prevent the progression of cancer. Consequently, translational medicines have been developed using metabolic substrates, precursors, and other products depending on their biochemical mechanism of action. It is important to note that these metabolic analogs can also be used for imaging and therapeutic purposes in addition to competing for metabolic functions. In particular, due to their isotopic labeling, these compounds may also be used to localize and visualize tumor cells after uptake. In this review, the current development status, applicability, and limitations of compounds targeting metabolic reprogramming are described, as well as the imaging platforms that are most suitable for each compound and the types of cancer to which they are most appropriate.
Collapse
Affiliation(s)
- Yi-Fang Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Huei-Yu Cai
- Department of Biomedicine Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei 11121, Taiwan
| | - Bo-Syuan Lin
- Department of Biomedicine Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei 11121, Taiwan
| | - Cheorl-Ho Kim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Seoburo 2066, Suwon 16419, Republic of Korea
- Samsung Advanced Institute of Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Yu-Chan Chang
- Department of Biomedicine Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei 11121, Taiwan
| |
Collapse
|
9
|
Jin C, Luo X, Li X, Zhou R, Zhong Y, Xu Z, Cui C, Xing X, Zhang H, Tian M. Positron emission tomography molecular imaging-based cancer phenotyping. Cancer 2022; 128:2704-2716. [PMID: 35417604 PMCID: PMC9324101 DOI: 10.1002/cncr.34228] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 12/28/2022]
Abstract
During the past several decades, numerous studies have provided insights into biological characteristics of cancer cells and identified various hallmarks of cancer acquired in the tumorigenic processes. However, it is still challenging to image these distinctive traits of cancer to facilitate the management of patients in clinical settings. The rapidly evolving field of positron emission tomography (PET) imaging has provided opportunities to investigate cancer's biological characteristics in vivo. This article reviews the current status of PET imaging on characterizing hallmarks of cancer and discusses the future directions of PET imaging strategies facilitating in vivo cancer phenotyping.
Collapse
Affiliation(s)
- Chentao Jin
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoyun Luo
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoyi Li
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Rui Zhou
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Yan Zhong
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Zhoujiao Xu
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Chunyi Cui
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoqing Xing
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Hong Zhang
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
- College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
- Key Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouChina
| | - Mei Tian
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
10
|
A robust [11C]acetate synthesis on a TRACERLab FX C pro module. Appl Radiat Isot 2022; 188:110356. [DOI: 10.1016/j.apradiso.2022.110356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/19/2022]
|
11
|
Liu M, Liu N, Wang J, Fu S, Wang X, Chen D. Acetyl-CoA Synthetase 2 as a Therapeutic Target in Tumor Metabolism. Cancers (Basel) 2022; 14:cancers14122896. [PMID: 35740562 PMCID: PMC9221533 DOI: 10.3390/cancers14122896] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/04/2022] [Accepted: 06/08/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Acetyl-CoA Synthetase 2 (ACSS2) is highly expressed in a variety of tumors, which is very important for tumor growth, proliferation, invasion, and metastasis in the nutritional stress microenvironment. Studies have proven that ACSS2 inhibitors can be effective in halting cancer growth and can be combined with other antineoplastic drugs to reduce drug resistance. This article mainly reviews the mechanism of ACSS2-promoting tumor growth from many aspects and the prospect of clinical application of targeted inhibitors. Abstract Acetyl-CoA Synthetase 2 (ACSS2) belongs to a member of the acyl-CoA short-chain synthase family, which can convert acetate in the cytoplasm and nucleus into acetyl-CoA. It has been proven that ACSS2 is highly expressed in glioblastoma, breast cancer, liver cancer, prostate cancer, bladder cancer, renal cancer, and other tumors, and is closely related to tumor stage and the overall survival rate of patients. Accumulating studies show that hypoxia and a low serum level induce ACSS2 expression to help tumor cells cope with this nutrient-poor environment. The potential mechanisms are associated with the ability of ACSS2 to promote the synthesis of lipids in the cytoplasm, induce the acetylation of histones in the nucleus, and facilitate the expression of autophagy genes. Novel-specific inhibitors of ACSS2 are developed and confirmed to the effectiveness in pre-clinical tumor models. Targeting ACSS2 may provide novel approaches for tumor treatment. This review summarizes the biological function of ACSS2, its relation to survival and prognosis in different tumors, and how ACSS2 mediates different pathways to promote tumor metastasis, invasion, and drug resistance.
Collapse
Affiliation(s)
| | | | | | | | - Xu Wang
- Correspondence: (X.W.); (D.C.)
| | | |
Collapse
|
12
|
Abstract
Renal cell carcinoma (RCC) is the sixth most common cancer among men and the ninth among women, and its prognosis is closely correlated with metastasis. Targeted therapy and immunotherapy are the main adjuvant treatments for advanced RCC and require early diagnosis, precise assessment, and prediction of the therapeutic responses. Current conventional imaging methods of RCC only provide structural information rather than biological processes. Noninvasive diagnostic tools are therefore needed to image RCC early and accurately at the molecular level. Nuclear medicine imaging combines the high sensitivity of radionuclides with the high resolution of structural imaging to visualize the metabolic processes and specific targets of RCC for more accurate and reliable diagnosis, staging, prognosis prediction, and response assessment. This review summarizes the most recent applications of nuclear medicine receptor imaging and metabolic imaging in RCC and highlights future development perspectives in the field.
Collapse
Affiliation(s)
- Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| |
Collapse
|
13
|
Crișan G, Moldovean-Cioroianu NS, Timaru DG, Andrieș G, Căinap C, Chiș V. Radiopharmaceuticals for PET and SPECT Imaging: A Literature Review over the Last Decade. Int J Mol Sci 2022; 23:5023. [PMID: 35563414 PMCID: PMC9103893 DOI: 10.3390/ijms23095023] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Positron emission tomography (PET) uses radioactive tracers and enables the functional imaging of several metabolic processes, blood flow measurements, regional chemical composition, and/or chemical absorption. Depending on the targeted processes within the living organism, different tracers are used for various medical conditions, such as cancer, particular brain pathologies, cardiac events, and bone lesions, where the most commonly used tracers are radiolabeled with 18F (e.g., [18F]-FDG and NA [18F]). Oxygen-15 isotope is mostly involved in blood flow measurements, whereas a wide array of 11C-based compounds have also been developed for neuronal disorders according to the affected neuroreceptors, prostate cancer, and lung carcinomas. In contrast, the single-photon emission computed tomography (SPECT) technique uses gamma-emitting radioisotopes and can be used to diagnose strokes, seizures, bone illnesses, and infections by gauging the blood flow and radio distribution within tissues and organs. The radioisotopes typically used in SPECT imaging are iodine-123, technetium-99m, xenon-133, thallium-201, and indium-111. This systematic review article aims to clarify and disseminate the available scientific literature focused on PET/SPECT radiotracers and to provide an overview of the conducted research within the past decade, with an additional focus on the novel radiopharmaceuticals developed for medical imaging.
Collapse
Affiliation(s)
- George Crișan
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | | | - Diana-Gabriela Timaru
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
| | - Gabriel Andrieș
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | - Călin Căinap
- The Oncology Institute “Prof. Dr. Ion Chiricuţă”, Republicii 34-36, 400015 Cluj-Napoca, Romania;
| | - Vasile Chiș
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Institute for Research, Development and Innovation in Applied Natural Sciences, Babeș-Bolyai University, Str. Fântânele 30, 400327 Cluj-Napoca, Romania
| |
Collapse
|
14
|
Abstract
The increasing number of different novel positron emission tomography (PET) radiopharmaceuticals poses challenges for their manufacturing procedures at different PET research facilities. Recent commercially available radiochemistry units with disposable cassettes are becoming common stations to produce radiopharmaceuticals with high specifications to understand the critical PET imaging outputs of the study. Therefore, several radiochemists across the PET research centers develop and optimize their own radiochemistry protocols to develop a novel or routine radiopharmaceutical at their lab. In this report, we describe the general procedure and steps followed to develop a (clinical-grade) radiopharmaceutical on a commercially available radiochemistry unit, TRASIS AIO. As an example, we use our routine protocol followed for the production of [11C]acetate, a fatty acid metabolic PET imaging ligand for several cancer imaging studies.
Collapse
Affiliation(s)
- Naresh Damuka
- Department of Radiology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | | |
Collapse
|
15
|
Gonçalves AC, Richiardone E, Jorge J, Polónia B, Xavier CPR, Salaroglio IC, Riganti C, Vasconcelos MH, Corbet C, Sarmento-Ribeiro AB. Impact of cancer metabolism on therapy resistance - Clinical implications. Drug Resist Updat 2021; 59:100797. [PMID: 34955385 DOI: 10.1016/j.drup.2021.100797] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite an increasing arsenal of anticancer therapies, many patients continue to have poor outcomes due to the therapeutic failures and tumor relapses. Indeed, the clinical efficacy of anticancer therapies is markedly limited by intrinsic and/or acquired resistance mechanisms that can occur in any tumor type and with any treatment. Thus, there is an urgent clinical need to implement fundamental changes in the tumor treatment paradigm by the development of new experimental strategies that can help to predict the occurrence of clinical drug resistance and to identify alternative therapeutic options. Apart from mutation-driven resistance mechanisms, tumor microenvironment (TME) conditions generate an intratumoral phenotypic heterogeneity that supports disease progression and dismal outcomes. Tumor cell metabolism is a prototypical example of dynamic, heterogeneous, and adaptive phenotypic trait, resulting from the combination of intrinsic [(epi)genetic changes, tissue of origin and differentiation dependency] and extrinsic (oxygen and nutrient availability, metabolic interactions within the TME) factors, enabling cancer cells to survive, metastasize and develop resistance to anticancer therapies. In this review, we summarize the current knowledge regarding metabolism-based mechanisms conferring adaptive resistance to chemo-, radio-and immunotherapies as well as targeted therapies. Furthermore, we report the role of TME-mediated intratumoral metabolic heterogeneity in therapy resistance and how adaptations in amino acid, glucose, and lipid metabolism support the growth of therapy-resistant cancers and/or cellular subpopulations. We also report the intricate interplay between tumor signaling and metabolic pathways in cancer cells and discuss how manipulating key metabolic enzymes and/or providing dietary changes may help to eradicate relapse-sustaining cancer cells. Finally, in the current era of personalized medicine, we describe the strategies that may be applied to implement metabolic profiling for tumor imaging, biomarker identification, selection of tailored treatments and monitoring therapy response during the clinical management of cancer patients.
Collapse
Affiliation(s)
- Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium
| | - Joana Jorge
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Bárbara Polónia
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | | | - Chiara Riganti
- Department of Oncology, School of Medicine, University of Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium.
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Hematology Service, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal.
| |
Collapse
|
16
|
Zhang Z, Liu S, Ma H, Xiang X, Nie D, Hu P, Tang G. Propionic Acid-Based PET Imaging of Prostate Cancer. Mol Imaging Biol 2021; 23:836-845. [PMID: 33876336 DOI: 10.1007/s11307-021-01608-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE This study aimed to evaluate the potential value of 2-[18F]fluoropropionic acid ([18F]FPA) for PET imaging of prostate cancer (PCa) and to explore the relationship between [18F]FPA accumulation and fatty acid synthase (FASN) levels in PCa models. The results of the first [18F]FPA PET study of a PCa patient are reported. PROCEDURES The LNCaP, PC-3 cell lines with high FASN expression, and DU145 cell lines with low FASN expression were selected for cell culture. A PET imaging comparison of [18F]FDG and [18F]FPA was performed in LNCaP, PC-3, and DU145 tumors. Additionally, in vivo inhibition experiments in those models were conducted with orlistat. In a human PET study, a patient with PCa before surgery was examined with [18F]FPA PET and [18F]FDG PET. RESULTS The uptake of [18F]FPA in the LNCaP and PC-3 tumors was higher than that of [18F]FDG (P<0.05 and P<0.05), but was lower in DU145 tumors (P<0.05). The accumulation (% ID/g) of [18F]FPA in the LNCaP, PC-3, and DU145 tumors decreased by 27.6, 40.5, and 11.7 %, respectively, after treatment with orlistat. The [18F]FPA showed higher radioactive uptake than [18F]FDG in the first PCa patient. CONCLUSIONS The [18F]FPA uptake in PCa models may be varies with fatty acid synthase activity and could be reduced after administration of a single FASN inhibitor, albeit the activity that is not measured directly. The [18F]FPA seems to be a potential broad-spectrum PET imaging agent and may serve as a valuable tool in the diagnosis of PCa in humans.
Collapse
Affiliation(s)
- Zhanwen Zhang
- Department of Nuclear Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shaoyu Liu
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Nuclear Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hui Ma
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xianhong Xiang
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dahong Nie
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ping Hu
- Department of Nuclear Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
| | - Ganghua Tang
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- Nanfang PET Center and Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
17
|
Chen M, Zhu W, Du J, Yang C, Han B, Zhou D, Huo L, Zhuang J. 11C-acetate positron emission tomography is more precise than 18F-fluorodeoxyglucose positron emission tomography in evaluating tumor burden and predicting disease risk of multiple myeloma. Sci Rep 2021; 11:22188. [PMID: 34773054 PMCID: PMC8590058 DOI: 10.1038/s41598-021-01740-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/17/2021] [Indexed: 11/25/2022] Open
Abstract
The optimal method of tumor burden evaluation in newly diagnosed multiple myeloma (NDMM) is yet to be determined. This study aimed to compare the value of 11C-acetate positron-emission tomography (PET)/computed tomography (CT) (AC-PET and 18F-fluorodeoxyglucose PET/CT (FDG-PET) in the assessment of tumor burden in NDMM.
This study evaluated 64 NDMM patients between February 2015 and July 2018. AC-PET and FDG-PET were used to assess myeloma lesions. The clinical data, imaging results, and their correlations were analyzed. Diffuse bone marrow uptake in AC-PET was significantly correlated with biomarkers for tumor burden, including serum hemoglobin (P = 0.020), M protein (P = 0.054), the percentage of bone marrow plasma cells (P < 0.001), and the Durie–Salmon stage of the disease (P = 0.007). The maximum standard uptake value (SUVmax) of focal lesions and high diffuse bone marrow uptake in AC-PET showed stronger correlations with high-risk disease (P = 0.017, P = 0.013) than those in FDG-PET. Moreover, the presence of diffuse bone marrow uptake, more than ten focal lesions, and an SUVmax of focal lesions of > 6.0 in AC-PET, but not in FDG-PET, predicted a higher probability of disease progression and shorter progression-free survival (P < 0.05). AC-PET outperformed FDG-PET in tumor burden evaluation and disease progression prediction in NDMM.
Collapse
Affiliation(s)
- Miao Chen
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Wenjia Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jianhua Du
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chen Yang
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Bing Han
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Daobin Zhou
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Junling Zhuang
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
18
|
Manafi-Farid R, Ranjbar S, Jamshidi Araghi Z, Pilz J, Schweighofer-Zwink G, Pirich C, Beheshti M. Molecular Imaging in Primary Staging of Prostate Cancer Patients: Current Aspects and Future Trends. Cancers (Basel) 2021; 13:5360. [PMID: 34771523 PMCID: PMC8582501 DOI: 10.3390/cancers13215360] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 12/19/2022] Open
Abstract
Accurate primary staging is the cornerstone in all malignancies. Different morphological imaging modalities are employed in the evaluation of prostate cancer (PCa). Regardless of all developments in imaging, invasive histopathologic evaluation is still the standard method for the detection and staging of the primary PCa. Magnetic resonance imaging (MRI) and computed tomography (CT) play crucial roles; however, functional imaging provides additional valuable information, and it is gaining ever-growing acceptance in the management of PCa. Targeted imaging with different radiotracers has remarkably evolved in the past two decades. [111In]In-capromab pendetide scintigraphy was a new approach in the management of PCa. Afterwards, positron emission tomography (PET) tracers such as [11C/18F]choline and [11C]acetate were developed. Nevertheless, none found a role in the primary staging. By introduction of the highly sensitive small molecule prostate-specific membrane antigen (PSMA) PET/CT, as well as recent developments in MRI and hybrid PET/MRI systems, non-invasive staging of PCa is being contemplated. Several studies investigated the role of these sophisticated modalities in the primary staging of PCa, showing promising results. Here, we recapitulate the role of targeted functional imaging. We briefly mention the most popular radiotracers, their diagnostic accuracy in the primary staging of PCa, and impact on patient management.
Collapse
Affiliation(s)
- Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran 1411713135, Iran;
| | - Shaghayegh Ranjbar
- Department of Nuclear Medicine, Division of Molecular Imaging and Theranostics, University Hospital Salzburg, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020 Salzburg, Austria; (S.R.); (Z.J.A.); (J.P.); (G.S.-Z.); (C.P.)
| | - Zahra Jamshidi Araghi
- Department of Nuclear Medicine, Division of Molecular Imaging and Theranostics, University Hospital Salzburg, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020 Salzburg, Austria; (S.R.); (Z.J.A.); (J.P.); (G.S.-Z.); (C.P.)
| | - Julia Pilz
- Department of Nuclear Medicine, Division of Molecular Imaging and Theranostics, University Hospital Salzburg, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020 Salzburg, Austria; (S.R.); (Z.J.A.); (J.P.); (G.S.-Z.); (C.P.)
| | - Gregor Schweighofer-Zwink
- Department of Nuclear Medicine, Division of Molecular Imaging and Theranostics, University Hospital Salzburg, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020 Salzburg, Austria; (S.R.); (Z.J.A.); (J.P.); (G.S.-Z.); (C.P.)
| | - Christian Pirich
- Department of Nuclear Medicine, Division of Molecular Imaging and Theranostics, University Hospital Salzburg, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020 Salzburg, Austria; (S.R.); (Z.J.A.); (J.P.); (G.S.-Z.); (C.P.)
| | - Mohsen Beheshti
- Department of Nuclear Medicine, Division of Molecular Imaging and Theranostics, University Hospital Salzburg, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020 Salzburg, Austria; (S.R.); (Z.J.A.); (J.P.); (G.S.-Z.); (C.P.)
| |
Collapse
|
19
|
Mosier JA, Schwager SC, Boyajian DA, Reinhart-King CA. Cancer cell metabolic plasticity in migration and metastasis. Clin Exp Metastasis 2021; 38:343-359. [PMID: 34076787 DOI: 10.1007/s10585-021-10102-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 05/08/2021] [Indexed: 12/13/2022]
Abstract
Metabolic reprogramming is a hallmark of cancer metastasis in which cancer cells manipulate their metabolic profile to meet the dynamic energetic requirements of the tumor microenvironment. Though cancer cell proliferation and migration through the extracellular matrix are key steps of cancer progression, they are not necessarily fueled by the same metabolites and energy production pathways. The two main metabolic pathways cancer cells use to derive energy from glucose, glycolysis and oxidative phosphorylation, are preferentially and plastically utilized by cancer cells depending on both their intrinsic metabolic properties and their surrounding environment. Mechanical factors in the microenvironment, such as collagen density, pore size, and alignment, and biochemical factors, such as oxygen and glucose availability, have been shown to influence both cell migration and glucose metabolism. As cancer cells have been identified as preferentially utilizing glycolysis or oxidative phosphorylation based on heterogeneous intrinsic or extrinsic factors, the relationship between cancer cell metabolism and metastatic potential is of recent interest. Here, we review current in vitro and in vivo findings in the context of cancer cell metabolism during migration and metastasis and extrapolate potential clinical applications of this work that could aid in diagnosing and tracking cancer progression in vivo by monitoring metabolism. We also review current progress in the development of a variety of metabolically targeted anti-metastatic drugs, both in clinical trials and approved for distribution, and highlight potential routes for incorporating our recent understanding of metabolic plasticity into therapeutic directions. By further understanding cancer cell energy production pathways and metabolic plasticity, more effective and successful clinical imaging and therapeutics can be developed to diagnose, target, and inhibit metastasis.
Collapse
Affiliation(s)
- Jenna A Mosier
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - David A Boyajian
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | | |
Collapse
|
20
|
Mirzaei R, Afaghi A, Babakhani S, Sohrabi MR, Hosseini-Fard SR, Babolhavaeji K, Khani Ali Akbari S, Yousefimashouf R, Karampoor S. Role of microbiota-derived short-chain fatty acids in cancer development and prevention. Biomed Pharmacother 2021; 139:111619. [PMID: 33906079 DOI: 10.1016/j.biopha.2021.111619] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/01/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Following cancer, cells in a particular tissue can no longer respond to the factors involved in controlling cell survival, differentiation, proliferation, and death. In recent years, it has been indicated that alterations in the gut microbiota components, intestinal epithelium, and host immune system are associated with cancer incidence. Also, it has been demonstrated that the short-chain fatty acids (SCFAs) generated by gut microbiota are vitally crucial in cell homeostasis as they contribute to the modulation of histone deacetylases (HDACs), resulting effected cell attachment, immune cell immigration, cytokine production, chemotaxis, and the programmed cell death. Therefore, the manipulation of SCFA levels in the intestinal tract by alterations in the microbiota structure can be potentially taken into consideration for cancer treatment/prevention. In the current study, we will explain the most recent findings on the detrimental or protective roles of SFCA (particularly butyrate, propionate, and acetate) in several cancers, including bladder, colon, breast, stomach, liver, lung, pancreas, and prostate cancers.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Azam Afaghi
- Department of Biology, Sofian Branch, Islamic Azad University, Sofian, Iran
| | - Sajad Babakhani
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Masoud Reza Sohrabi
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiandokht Babolhavaeji
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shabnam Khani Ali Akbari
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Miller KD, Pniewski K, Perry CE, Papp SB, Shaffer JD, Velasco-Silva JN, Casciano JC, Aramburu TM, Srikanth YVV, Cassel J, Skordalakes E, Kossenkov AV, Salvino JM, Schug ZT. Targeting ACSS2 with a Transition-State Mimetic Inhibits Triple-Negative Breast Cancer Growth. Cancer Res 2021; 81:1252-1264. [PMID: 33414169 PMCID: PMC8026699 DOI: 10.1158/0008-5472.can-20-1847] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/20/2020] [Accepted: 12/29/2020] [Indexed: 11/16/2022]
Abstract
Acetyl-CoA is a vitally important and versatile metabolite used for many cellular processes including fatty acid synthesis, ATP production, and protein acetylation. Recent studies have shown that cancer cells upregulate acetyl-CoA synthetase 2 (ACSS2), an enzyme that converts acetate to acetyl-CoA, in response to stresses such as low nutrient availability and hypoxia. Stressed cancer cells use ACSS2 as a means to exploit acetate as an alternative nutrient source. Genetic depletion of ACSS2 in tumors inhibits the growth of a wide variety of cancers. However, there are no studies on the use of an ACSS2 inhibitor to block tumor growth. In this study, we synthesized a small-molecule inhibitor that acts as a transition-state mimetic to block ACSS2 activity in vitro and in vivo. Pharmacologic inhibition of ACSS2 as a single agent impaired breast tumor growth. Collectively, our findings suggest that targeting ACSS2 may be an effective therapeutic approach for the treatment of patients with breast cancer. SIGNIFICANCE: These findings suggest that targeting acetate metabolism through ACSS2 inhibitors has the potential to safely and effectively treat a wide range of patients with cancer.
Collapse
Affiliation(s)
- Katelyn D Miller
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania
| | - Katherine Pniewski
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania
| | - Caroline E Perry
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania
- Cell & Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sara B Papp
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania
| | - Joshua D Shaffer
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania
- Cell & Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jesse N Velasco-Silva
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania
- Biochemistry Department, School of Medicine, University of Utah, Salt Lake City, Utah
| | - Jessica C Casciano
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania
| | - Tomas M Aramburu
- Gene Expression and Regulation Program, Wistar Institute, Philadelphia, Pennsylvania
| | | | - Joel Cassel
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania
| | - Emmanuel Skordalakes
- Gene Expression and Regulation Program, Wistar Institute, Philadelphia, Pennsylvania
| | - Andrew V Kossenkov
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania
| | - Joseph M Salvino
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania
| | - Zachary T Schug
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, Pennsylvania.
| |
Collapse
|
22
|
Bader DA, McGuire SE. Tumour metabolism and its unique properties in prostate adenocarcinoma. Nat Rev Urol 2020; 17:214-231. [PMID: 32112053 DOI: 10.1038/s41585-020-0288-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2020] [Indexed: 12/14/2022]
Abstract
Anabolic metabolism mediated by aberrant growth factor signalling fuels tumour growth and progression. The first biochemical descriptions of the altered metabolic nature of solid tumours were reported by Otto Warburg almost a century ago. Now, the study of tumour metabolism is being redefined by the development of new molecular tools, tumour modelling systems and precise instrumentation together with important advances in genetics, cell biology and spectroscopy. In contrast to Warburg's original hypothesis, accumulating evidence demonstrates a critical role for mitochondrial metabolism and substantial variation in the way in which different tumours metabolize nutrients to generate biomass. Furthermore, computational and experimental approaches suggest a dominant influence of the tissue-of-origin in shaping the metabolic reprogramming that enables tumour growth. For example, the unique metabolic properties of prostate adenocarcinoma are likely to stem from the distinct metabolism of the prostatic epithelium from which it emerges. Normal prostatic epithelium employs comparatively glycolytic metabolism to sustain physiological citrate secretion, whereas prostate adenocarcinoma consumes citrate to power oxidative phosphorylation and fuel lipogenesis, enabling tumour progression through metabolic reprogramming. Current data suggest that the distinct metabolic aberrations in prostate adenocarcinoma are driven by the androgen receptor, providing opportunities for functional metabolic imaging and novel therapeutic interventions that will be complementary to existing diagnostic and treatment options.
Collapse
Affiliation(s)
- David A Bader
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA. .,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA.
| | - Sean E McGuire
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA. .,Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Calabria F, Pichler R, Leporace M, Wolfsgruber J, Coscarelli P, Dunzinger A, Schillaci O, Cascini GL, Bagnato A. 68Ga/64Cu PSMA Bio-Distribution in Prostate Cancer Patients: Potential Pitfalls for Different Tracers. Curr Radiopharm 2020; 12:238-246. [PMID: 31113354 DOI: 10.2174/1874471012666190515090755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 03/20/2019] [Accepted: 04/04/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND 68Ga-PSMA is a widely useful PET/CT tracer for prostate cancer imaging. Being a transmembrane protein acting as a glutamate carboxypeptidase enzyme, PSMA is highly expressed in prostate cancer cells. PSMA can also be labeled with 64Cu, offering a longer half-life and different resolution imaging. Several studies documented bio-distribution and pitfalls of 68Ga-PSMA as well as of 64Cu- PSMA. No data are reported on differences between these two variants of PSMA. Our aim was to evaluate physiological distribution of these two tracers and to analyze false positive cases. METHODS We examined tracer bio-distribution in prostate cancer patients with negative 68Ga-PSMA PET/CT (n=20) and negative 64Ga-PSMA PET/CT (n=10). A diagnostic pitfall for each tracer was documented. RESULT Bio-distribution of both tracers was similar, with some differences due to renal excretion of 68Ga- PSMA and biliary excretion of 64Cu-PSMA. 68Ga-PSMA uptake was observed in sarcoidosis while 64Cu- PSMA uptake was recorded in pneumonitis. DISCUSSION Both tracers may present similar bio-distribution in the human body, with similar uptake in exocrine glands and high intestinal uptake. Similarly to other tracers, false positive cases cannot be excluded in clinical practice. CONCLUSION The knowledge of difference in bio-distribution between two tracers may help in interpretation of PET data. Diagnostic pitfalls can be documented, due to the possibility of PSMA uptake in inflammation. Our results are preliminary to future studies comparing diagnostic accuracies of 68Ga-PSMA and 64Cu-PSMA.
Collapse
Affiliation(s)
- Ferdinando Calabria
- Department of Nuclear Medicine and Theranostics, National Public Hospital "Mariano Santo", 87100, Cosenza, Italy
| | - Robert Pichler
- Institute of Nuclear Medicine, Kepler University Hospital, Neuromed Campus, Wagner-Jauregg Weg 15, A-4021 Linz, Austria
| | - Mario Leporace
- Department of Nuclear Medicine and Theranostics, National Public Hospital "Mariano Santo", 87100, Cosenza, Italy
| | | | | | - Andreas Dunzinger
- Institute of Nuclear Medicine, Kepler University Hospital, Neuromed Campus, Wagner-Jauregg Weg 15, A-4021 Linz, Austria
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University "Tor Vergata", 00133, Rome, Italy.,IRCCS INM Neuromed, 86077, Pozzilli (IS), Italy
| | - Giuseppe Lucio Cascini
- Department of Diagnostic Imaging, Nuclear Medicine Unit, Magna Graecia University, Catanzaro, Italy
| | - Antonio Bagnato
- Department of Nuclear Medicine and Theranostics, National Public Hospital "Mariano Santo", 87100, Cosenza, Italy
| |
Collapse
|
24
|
Regula N, Honarvar H, Lubberink M, Jorulf H, Ladjevardi S, Häggman M, Antoni G, Buijs J, Velikyan I, Sörensen J. Carbon Flux as a Measure of Prostate Cancer Aggressiveness: [ 11C]-Acetate PET/CT. Int J Med Sci 2020; 17:214-223. [PMID: 32038105 PMCID: PMC6990881 DOI: 10.7150/ijms.39542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/12/2019] [Indexed: 01/15/2023] Open
Abstract
Purpose: Dynamic [11C]-acetate positron emission tomography (PET) can be used to study tissue perfusion and carbon flux simultaneously. In this study, the feasibility of the quantification of prostate cancer aggressiveness using parametric methods assessing [11C]-acetate kinetics was investigated in prostate cancer subjects. The underlying uptake mechanism correlated with [11C]-acetate influx and efflux measured in real-time in vitro in cell culture. Methods: Twenty-one patients with newly diagnosed low-to-moderate risk prostate cancer underwent magnetic resonance imaging (MRI) and dynamic [11C]-acetate PET/CT examinations of the pelvis. Parametric images of K1 (extraction × perfusion), k2 (oxidative metabolism) and VT (=K1/k2, anabolic metabolism defined as carbon retention) were constructed using a one-tissue compartment model with an arterial input function derived from pelvic arteries. Regions of interest (ROIs) of the largest cancer lesion in each patient and normal prostate tissue were drawn using information from MRI (T2 and DWI images), biopsy results, and post-surgical histopathology of whole prostate sections (n=7). In vitro kinetics of [11C]-acetate were studied on DU145 and PC3 cell lines using LigandTracer® White equipment for the measurement of the radioactivity uptake in real-time at 37°C. Results: Mean prostate specific antigen (PSA) was 8.33±3.92 ng/mL and median Gleason Sum 6 (range 5-7). K1, VT and standardized uptake values (SUVs) were significantly higher in cancerous prostate tissues compared to normal ones for all patients (p<0.001), while k2 was not (p=0.26). PSA values correlated to early SUVs (r=0.50, p=0.02) and K1 (r=0.48, p=0.03). Early and late SUVs correlated to VT (r>0.76, p<0.001) and K1 (r>0.64, p<0.005). In vitro studies demonstrated higher extraction and retention (p<0.01) of [11C]-acetate in the more aggressive PC3 cells. Conclusion: Parametric images could be used to visualize the [11C]-acetate kinetics of the prostate cancer exhibiting elevated extraction associated with the cancer aggressiveness. The influx rate of [11C]-acetate studied in cell culture also showed dependence on the cancer aggressiveness associated with elevated lipogenesis. Dynamic [11C]-acetate/PET demonstrated potential for prostate cancer aggressiveness estimation using parametric-based K1 and VT values.
Collapse
Affiliation(s)
- Naresh Regula
- Division of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Hadis Honarvar
- Division of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Mark Lubberink
- Division of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.,Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Håkan Jorulf
- Division of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Sam Ladjevardi
- Division of Urology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Michael Häggman
- Division of Urology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Gunnar Antoni
- Division of Molecular Imaging, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jos Buijs
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Irina Velikyan
- Division of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jens Sörensen
- Division of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.,PET Centre, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
25
|
Bispecific GRPR-Antagonistic Anti-PSMA/GRPR Heterodimer for PET and SPECT Diagnostic Imaging of Prostate Cancer. Cancers (Basel) 2019; 11:cancers11091371. [PMID: 31540122 PMCID: PMC6771040 DOI: 10.3390/cancers11091371] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 12/19/2022] Open
Abstract
Simultaneous targeting of the prostate-specific membrane antigen (PSMA) and gastrin-releasing peptide receptor (GRPR) could improve the diagnostic accuracy in prostate cancer (PCa). The aim of this study was to develop a PSMA/GRPR-targeting bispecific heterodimer for SPECT and positron emission tomography (PET) diagnostic imaging of PCa. The heterodimer NOTA-DUPA-RM26 was produced by manual solid-phase peptide synthesis. NOTA-DUPA-RM26 was labeled with 111In and 68Ga, with yields >98%, and demonstrated a high stability and binding specificity to PSMA and GRPR. IC50 values for natIn-NOTA-DUPA-RM26 were 4 ± 1 nM towards GRPR and 824 ± 230 nM towards PSMA. An in vivo binding specificity 1 h pi of 111In-NOTA-DUPA-RM26 in PC3-PIP-xenografted mice demonstrated partially blockable tumor uptake when co-injected with an excess of PSMA- or GRPR-targeting agents. Simultaneous co-injection of both agents induced pronounced blocking. The biodistribution of 111In-NOTA-DUPA-RM26 and 68Ga-NOTA-DUPA-RM26 revealed fast activity clearance from the blood and normal organs via the kidneys. Tumor uptake exceeded normal organ uptake for both analogs 1 h pi. 68Ga-NOTA-DUPA-RM26 had a significantly lower tumor uptake (8 ± 2%ID/g) compared to 111In-NOTA-DUPA-RM26 (12 ± 2%ID/g) 1 h pi. Tumor-to-organ ratios increased 3 h pi, but decreased 24 h pi, for 111In-NOTA-DUPA-RM26. MicroPET/CT and microSPECT/CT scans confirmed biodistribution data, suggesting that 68Ga-NOTA-DUPA-RM26 and 111In-NOTA-DUPA-RM26 are suitable candidates for the imaging of GRPR and PSMA expression in PCa shortly after administration.
Collapse
|
26
|
Schollhammer R, De Clermont Gallerande H, Yacoub M, Quintyn Ranty ML, Barthe N, Vimont D, Hindié E, Fernandez P, Morgat C. Comparison of the radiolabeled PSMA-inhibitor 111In-PSMA-617 and the radiolabeled GRP-R antagonist 111In-RM2 in primary prostate cancer samples. EJNMMI Res 2019; 9:52. [PMID: 31161459 PMCID: PMC6546761 DOI: 10.1186/s13550-019-0517-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose Prostate-specific membrane antigen (PSMA) and gastrin-releasing peptide receptor (GRP-R) are expressed in prostate cancer and can be targeted with radiolabeled inhibitors and antagonists. Their performances for the initial characterization of prostatic tumors have been barely evaluated but never compared. We aimed to gather comparative preclinical data of the role of PSMA and GRP-R targeting in prostate cancer. Procedures We retrospectively studied 20 frozen prostatectomy samples with various metastatic risks of the D’Amico classification. Tissue samples were investigated by tissular microimaging using the radiolabeled PSMA inhibitor 111In-PSMA-617 and the radiolabeled GRP-R antagonist 111In-RM2. Bindings of the two radiopharmaceuticals were compared to histology and clinico-biological data (Gleason score, PSA values, metastatic risks). Results Binding of 111In-PSMA-617 was high whatever the metastatic risk (p = 0.665), Gleason score (p = 0.555), or PSA value (p = 0.404) while 111In-RM2 exhibited a significantly higher binding in the low metastatic risk group (p = 0.046), in the low PSA value group (p = 0.001), and in samples with Gleason 6 score (p = 0.006). Conclusion PSMA and GRP-R based imaging might have complementary performances for the initial characterization of prostatic tumors. Prospective clinical studies comparing the two tracers in this setting are needed.
Collapse
Affiliation(s)
- Romain Schollhammer
- Nuclear Medicine Department, University Hospital of Bordeaux, Place Amélie Raba Léon, 33000, 33076, Bordeaux, France. .,University of Bordeaux, INCIA, UMR5287, 33400, Talence, France. .,CNRS, INCIA, UMR5287, 33400, Talence, France.
| | - Henri De Clermont Gallerande
- Nuclear Medicine Department, University Hospital of Bordeaux, Place Amélie Raba Léon, 33000, 33076, Bordeaux, France
| | - Mokrane Yacoub
- Department of Pathology, University Hospital of Bordeaux, 33076, Bordeaux, France
| | | | | | - Delphine Vimont
- University of Bordeaux, INCIA, UMR5287, 33400, Talence, France.,CNRS, INCIA, UMR5287, 33400, Talence, France
| | - Elif Hindié
- Nuclear Medicine Department, University Hospital of Bordeaux, Place Amélie Raba Léon, 33000, 33076, Bordeaux, France.,University of Bordeaux, INCIA, UMR5287, 33400, Talence, France.,CNRS, INCIA, UMR5287, 33400, Talence, France
| | - Philippe Fernandez
- Nuclear Medicine Department, University Hospital of Bordeaux, Place Amélie Raba Léon, 33000, 33076, Bordeaux, France.,University of Bordeaux, INCIA, UMR5287, 33400, Talence, France.,CNRS, INCIA, UMR5287, 33400, Talence, France
| | - Clément Morgat
- Nuclear Medicine Department, University Hospital of Bordeaux, Place Amélie Raba Léon, 33000, 33076, Bordeaux, France.,University of Bordeaux, INCIA, UMR5287, 33400, Talence, France.,CNRS, INCIA, UMR5287, 33400, Talence, France
| |
Collapse
|
27
|
Xu KM, Chen RC, Schuster DM, Jani AB. Role of novel imaging in the management of prostate cancer. Urol Oncol 2019; 37:611-618. [PMID: 31072791 DOI: 10.1016/j.urolonc.2019.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 03/13/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022]
Abstract
This review summarizes novel imaging in the management of prostate cancer including multiparametric MRI, PET-CT scans with different radiotracers including 11C-acetate, 11C-choline, 18F-choline, 18F sodium fluoride, prostate-specific membrane antigen, and anti-1-amino-3-[18F] fluorocyclobutane-1-carboxylic acid (fluciclovine).
Collapse
Affiliation(s)
- Karen M Xu
- Department of Radiation Oncology and Winship Cancer Institute of Emory University, Emory University, Atlanta, GA
| | - Ronald C Chen
- Department of Radiation Oncology, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - David M Schuster
- Department of Radiology and Imaging Sciences, Division of Nuclear Medicine and Molecular Imaging, Emory University Hospital, Atlanta, GA
| | - Ashesh B Jani
- Department of Radiation Oncology and Winship Cancer Institute of Emory University, Emory University, Atlanta, GA.
| |
Collapse
|
28
|
Zadra G, Loda M. When fat goes down, prostate cancer is on the ropes. Mol Cell Oncol 2019; 6:1595308. [PMID: 31131311 PMCID: PMC6512936 DOI: 10.1080/23723556.2019.1595308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/10/2019] [Accepted: 03/12/2019] [Indexed: 01/28/2023]
Abstract
Reprogrammed lipid metabolism and persistent androgen receptor signaling commonly mark aggressive prostate cancer. We describe that targeting de-novo lipogenesis deprives prostate cancer cells of substrates and fuel, while inhibiting androgen receptor signaling. Our study uncovers the interplay between lipogenesis and androgen receptor and proposes novel combinatorial therapeutic approaches.
Collapse
Affiliation(s)
- Giorgia Zadra
- Dana-Farber Cancer Institute, Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Massimo Loda
- Dana-Farber Cancer Institute, Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA.,Broad Institute, Cambridge, MA, USA.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NY, NY, USA
| |
Collapse
|
29
|
Liu C, Xing M, Cong B, Qiu C, He D, Wang C, Xiao Y, Yin T, Shao M, Qiu W, Ma T, Gong X, Chen X, Zheng H, Zheng R, Song L. In vivo transrectal imaging of canine prostate with a sensitive and compact handheld transrectal array photoacoustic probe for early diagnosis of prostate cancer. BIOMEDICAL OPTICS EXPRESS 2019; 10:1707-1717. [PMID: 31086699 PMCID: PMC6484995 DOI: 10.1364/boe.10.001707] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/28/2019] [Accepted: 02/17/2019] [Indexed: 05/10/2023]
Abstract
In this study, we built a novel handheld array photoacoustic probe by integrating multiple optical components with a transrectal ultrasound (TRUS) transducer array. The optical components deliver laser energy and TRUS is used for photoacoustic signal detection. Compared to the previously reported probe, the new photoacoustic probe utilizes an optimized light illumination scheme to enhance the utilization-efficiency of the laser energy, thus improving the imaging sensitivity of the probe. In addition, the new probe is compact and easy to handle for clinicians. We validated the use of this photoacoustic probe for prostate cancer imaging through both phantom studies and in vivo canine model study, which mimics the prostate cancer conditions. The results showed that the probe is suitable for clinical use and can be used in the clinics for several potential clinical applications, including early diagnosis of prostate cancer, targeted image-guided biopsy, and image-guided intra-operative procedure.
Collapse
Affiliation(s)
- Chengbo Liu
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Shenzhen 518055, China
| | - Muyue Xing
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Bing Cong
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Chen Qiu
- Department of Medical Ultrasound, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Dong He
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Congzhi Wang
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Shenzhen 518055, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yang Xiao
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Shenzhen 518055, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tinghui Yin
- Department of Medical Ultrasound, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Min Shao
- SonoScape Corporation, Shenzhen 518051, China
| | - Weibao Qiu
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Shenzhen 518055, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Teng Ma
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Shenzhen 518055, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaojing Gong
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Shenzhen 518055, China
| | - Xiong Chen
- SonoScape Corporation, Shenzhen 518051, China
| | - Hairong Zheng
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Shenzhen 518055, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Rongqin Zheng
- Department of Medical Ultrasound, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Liang Song
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Shenzhen 518055, China
| |
Collapse
|
30
|
Graham K, Unger E. Overcoming tumor hypoxia as a barrier to radiotherapy, chemotherapy and immunotherapy in cancer treatment. Int J Nanomedicine 2018; 13:6049-6058. [PMID: 30323592 PMCID: PMC6177375 DOI: 10.2147/ijn.s140462] [Citation(s) in RCA: 391] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypoxia exists to some degree in most solid tumors due to inadequate oxygen delivery of the abnormal vasculature which cannot meet the demands of the rapidly proliferating cancer cells. The levels of oxygenation within the same tumor are highly variable from one area to another and can change over time. Tumor hypoxia is an important impediment to effective cancer therapy. In radiotherapy, the primary mechanism is the creation of reactive oxygen species; hypoxic tumors are therefore radiation resistant. A number of chemotherapeutic drugs have been shown to be less effective when exposed to a hypoxic environment which can lead to further disease progression. Hypoxia is also a potent barrier to effective immunotherapy in cancer treatment. Because of the recognition of hypoxia as an important barrier to cancer treatment, a variety of approaches have been undertaken to overcome or reverse tumor hypoxia. Such approaches have included breathing hyperbaric oxygen, artificial hemoglobins, allosteric hemoglobin modifiers, hypoxia activated prodrugs and fluorocarbons (FCs). These approaches have largely failed due to limited efficacy and/or adverse side effects. Oxygen therapeutics, based on liquid FCs, can potentially increase the oxygen-carrying capacity of the blood to reverse tumor hypoxia. Currently, at least two drugs are in clinical trials to reverse tumor hypoxia; one of these is designed to improve permeability of oxygen into the tumor tissue and the other is based upon a low boiling point FC that transports higher amounts of oxygen per gram than previously tested FCs.
Collapse
|
31
|
Pantel AR, Ackerman D, Lee SC, Mankoff DA, Gade TP. Imaging Cancer Metabolism: Underlying Biology and Emerging Strategies. J Nucl Med 2018; 59:1340-1349. [PMID: 30042161 PMCID: PMC6126440 DOI: 10.2967/jnumed.117.199869] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 06/18/2018] [Indexed: 12/22/2022] Open
Abstract
Dysregulated cellular metabolism is a characteristic feature of malignancy that has been exploited for both imaging and targeted therapy. With regard to imaging, deranged glucose metabolism has been leveraged using 18F-FDG PET. Metabolic imaging with 18F-FDG, however, probes only the early steps of glycolysis; the complexities of metabolism beyond these early steps in this single pathway are not directly captured. New imaging technologies-both PET with novel radiotracers and MR-based methods-provide unique opportunities to investigate other aspects of cellular metabolism and expand the metabolic imaging armamentarium. This review will discuss the underlying biology of metabolic dysregulation in cancer, focusing on glucose, glutamine, and acetate metabolism. Novel imaging strategies will be discussed within this biologic framework, highlighting particular strengths and limitations of each technique. Emphasis is placed on the role that combining modalities will play in enabling multiparametric imaging to fully characterize tumor biology to better inform treatment.
Collapse
Affiliation(s)
- Austin R Pantel
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel Ackerman
- Penn Image-Guided Interventions Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Seung-Cheol Lee
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David A Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Terence P Gade
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Penn Image-Guided Interventions Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
32
|
Giunchi F, Fiorentino M, Loda M. The Metabolic Landscape of Prostate Cancer. Eur Urol Oncol 2018; 2:28-36. [PMID: 30929843 DOI: 10.1016/j.euo.2018.06.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/30/2018] [Accepted: 06/22/2018] [Indexed: 12/28/2022]
Abstract
CONTEXT Neoplastic cells are characterized by metabolic alterations that sustain tumor growth. Interventions aimed at modifying metabolic rewiring of cancer cells are currently being investigated in several tumor types, including prostate cancer (PC). OBJECTIVE To review relevant metabolic alterations reported for PC and potential diagnostic and therapeutic opportunities that could be exploited on the basis of these discoveries. EVIDENCE ACQUISITION We performed a review of PubMed/Medline in March 2018 for PC in association with each of the following search terms: metabolomics; lipid, cholesterol, one-carbon, amino acid, and glucose metabolism. Fifty publications were selected for inclusion in this analysis. EVIDENCE SYNTHESIS The reports included were grouped according to fatty acid and cholesterol metabolism (28 studies); one-carbon metabolism (9 studies); amino acid metabolism (6 studies); and glucose metabolism (7 studies). We report on multiple metabolic pathways that are dysregulated in prostate cancer. Metabolic alterations can result in at least one of the following changes: protein lipidation, oncogene activation, DNA methylation, cellular signaling, and protein-protein interactions. CONCLUSIONS Metabolic alterations play a crucial role in PC development, progression, and resistance to therapy. Increasing knowledge of metabolic rewiring is revealing novel metabolic signatures in PC. These signatures could be utilized for PC diagnosis, as well as for the discovery of novel therapeutic interventions to overcome castration resistance. PATIENT SUMMARY Metabolic alterations play a crucial role in the development and progression of prostate cancer and its resistance to therapy. Our knowledge of metabolic rewiring is increasing and revealing novel metabolic signatures in prostate cancer. These signatures could be used for diagnosis and for the discovery of novel therapeutic interventions aimed at overcoming castration resistance.
Collapse
Affiliation(s)
- Francesca Giunchi
- Division of Genito-Urinary Pathology, S.Orsola-Malpighi Teaching Hospital, University of Bologna, Bologna, Italy
| | - Michelangelo Fiorentino
- Division of Genito-Urinary Pathology, S.Orsola-Malpighi Teaching Hospital, University of Bologna, Bologna, Italy.
| | - Massimo Loda
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Long-term Results of a Comparative PET/CT and PET/MRI Study of 11C-Acetate and 18F-Fluorocholine for Restaging of Early Recurrent Prostate Cancer. Clin Nucl Med 2017; 42:e242-e246. [PMID: 28240662 DOI: 10.1097/rlu.0000000000001609] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE The aims of this study were to assess the intraindividual performance of F-fluorocholine (FCH) and C-acetate (ACE) PET studies for restaging of recurrent prostate cancer (PCa), to correlate PET findings with long-term clinical and imaging follow-up, and to evaluate the impact of PET results on patient management. METHODS Thirty-three PCa patients relapsing after radical prostatectomy (n = 10, prostate-specific antigen [PSA] ≤3 ng/mL), primary radiotherapy (n = 8, prostate-specific antigen ≤5 ng/mL), or radical prostatectomy + salvage radiotherapy (n = 15) underwent ACE and FCH PET-CT (n = 29) or PET-MRI (n = 4) studies in a randomized sequence 0 to 21 days apart. RESULTS The detection rate for ACE was 66% and for FCH was 60%. Results were concordant in 79% of the cases (26/33) and discordant in 21% (retroperitoneal, n = 5; pararectal, n = 1; and external iliac nodes, n = 1). After a median FU of 41 months (n = 32, 1 patient lost to FU), the site of relapse was correctly identified by ACE and FCH in 53% (17/32) and 47% (15/32) of the patients, respectively (2 M1a patients ACE+/FCH-), whereas in 6 of 32 patients the relapse was not localized. Treatment approach was changed in 11 (34.4%) of 32 patients and 9 (28%) of 32 patients restaged with ACE and FCH PET, respectively. CONCLUSIONS In early recurrent PCa, ACE and FCH showed minor discrepancies, limited to nodal staging and mainly in the retroperitoneal area, with true positivity of PET findings confirmed in half of the cases during FU. Treatment approach turned out to be influenced by ACE or FCH PET studies in one third of the patients.
Collapse
|
34
|
Jadvar H. Radiotheranostics in Prostate Cancer: Introduction and Overview. J Nucl Med 2016; 57:1S-2S. [DOI: 10.2967/jnumed.116.183517] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/01/2016] [Indexed: 12/17/2022] Open
|